Galectin‑3 induces the phenotype transformation of human vascular smooth muscle cells via the canonical Wnt signaling

  • Authors:
    • Lei Tian
    • Kan Chen
    • Jiatian Cao
    • Zhihua Han
    • Yue Wang
    • Lin Gao
    • Yuqi Fan
    • Changqian Wang
  • View Affiliations

  • Published online on: April 3, 2017     https://doi.org/10.3892/mmr.2017.6429
  • Pages: 3840-3846
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Galectin‑3, a galactoside‑binding protein, is highly expressed in carotid plaques and plays an important role in the atherosclerotic lesions. The phenotype transformation of vascular smooth muscle cells is the basic pathological change of atherosclerosis. This study investigated the effects of exogenous galectin‑3 on the function and phenotype transformation of human umbilical vascular smooth muscle cells (HUSMC). In this study, we treated vascular smooth muscle cells with recombinant galectin‑3 and tested its effect on cell proliferation, migration, and phenotype transformation. Our results showed that exogenous galectin‑3 promoted human umbilical vascular smooth muscle cells (HUSMC) proliferation and migration. Exogenous galectin‑3 enhanced the expression of the smooth muscle synthetic protein osteopontin, smooth muscle contractile proteins calponin and smooth muscle α‑actin. The galectin‑3‑induced change in cell phenotype was associated with the activation of canonical Wnt signaling, as measured by β‑catenin axin2 and cyclin D1 expression. β‑catenin inhibition by small interfering RNA reduced cell proliferation, decreased cell motility, and blocked galectin‑3‑induced phenotype transformation of human umbilical vascular smooth muscle cells (HUSMC). Our data suggest galectin‑3 promotes the phenotype transformation of human umbilical vascular smooth muscle cells (HUSMC) by activating Wnt/β‑catenin signaling pathway.

Introduction

Atherosclerosis is the primary cause of heart disease and stroke, and it is also one of the main causes of death and disability around the world (1,2). The phenotype transformation of vascular smooth muscle cell (VSMC) plays an important role in the pathology process of atherosclerosis. In the atherosclerotic lesion, there is a large amount of the synthetic phenotype of VSMC in the walls of blood vessel. These synthetic phenotype VSMC tends to form foam-like cells and accelerate lesion progression (3,4). First, these cells secrete a variety of proteins (MMP-2, MMP-9, collagen type I and OPN), which increase lipid content and enhance the accumulation of monocytes and macrophages, second, they are believed to be activated because of enhancing ability of proliferation, immigration, and phagocytosis (3).

Galectin-3 (gal-3), a galactoside-binding protein, can be widely expressed by different kind of cells. Oxidized low-density lipoprotein (oxLDL) can also promote the expression of gal-3 in macrophages (5). In human umbilical vein endothelial cells (HUVEC), gal-3 plays an important role in vascular endothelial growth factor (VEGF)-and basic fibroblast growth factor (bFGF)-mediated angiogenesis (6). More importantly, Gal-3 is an important player in Aldo-induced vascular inflammation and mediates aldo-induced vascular fibrosis, silencing gal-3 blocks aldo-induced collagen type I deposition both in vivo and in vitro (7).

Gal-3 can regulate Wnt signaling (8). Gal-3 is a key regulator in the Wnt/β-catenin signaling pathway and interact with axin-2, GSK-3β and β-catenin immediately (911). Furthermore, gal-3 can in direct contact with β-catenin to stimulate cyclin D1 and c-myc expression (9). Tatsuo even find that human gal-3 sequence has a structural similarity to β-catenin in the breast cancer cells (10).

Recently, the role of gal-3 in cardiovascular disease has also been widely studied by some researchers (1217). In the setting of atherosclerotic disease, gal-3 seems to promote atherogenesis. Gal-3 is an useful biomarker which can predict the subsequent infarction after first myocardial infarction (MI) (18). Furthermore, in the absence of gal-3, the development of atherosclerotic pathological change in the ApoE-deficient mouse is reduced (19). Patients with type 2 diabetes and arterial hypertension have higher levels of gal-3 in plasma (20). Gal-3 has been proved to be a promising biomarker for detecting prediabetes and diabetes (21). Until now the mechanism of gal-3 in atherogenesis is still not clear yet, we presumed that gal-3 was a high risk factors of atherosclerosis and could promote pathological process of atherosclerosis. Considering the importance of phenotype transformation of SMC, does gal-3 induce atherosclerosis development by modulate cellular phenotype? If it does, what is the mechanism involved?

Thus, in this study, we tried to investigate the effects of exogenous gal-3 inHUSMCs. Furthermore, we examined whether β-catenin, a reported signaling associated with phenotype transformation of vascular smooth muscle cells, is involved in this gal-3 activity.

Materials and methods

Reagents

The Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), and penicillin/streptomycin (pen/strep, 10,000 U/ml each) were purchased from Gibco (Carlsbad, CA, USA). oxLDL was obtained from Peking Union-Biology Co., Ltd. (Beijing China). TRIzol reagent for RNA isolation was purchased from Invitrogen (Carlsbad, CA, USA). XAV939 was purchased from Sigma-Aldrich (St. Louis, MO, USA). The reverse transcriptase kit (RR037A) and SYBR Premix Ex Taq (DRR420S) were purchased from Takara (Dalian China). The Cell Counting Kit-8 (CCK-8) assay kit was purchased from Dojindo (Kumamoto, Japan). Recombinant human gal-3 was purchased from PeproTech (Rocky Hill, NJ, USA). The primary antibody against gal-3 (no. sc-20157), SMA (no. sc-32251), cyclin D1 (no. sc-8396) and GAPDH (no. sc-48166) were obtained from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Anti-phospho GSK3β (Ser9) (no. CST-9322), anti-GSK3β (no. CST-9315), anti-nonphospho (active) β-catenin (no. CST-4270), and anti-β-catenin (no. CST-9582) were acquired from Cell Signalling Technology, Inc. (Danvers, MA, USA). The antibody against osteopontin (OPN; no. ab91655), calponin (no. ab46794) and axin2 (no. ab109307) were obtained from Abcam (Cambridge, UK). The goat anti-rabbit secondary antibody (no. A-21109) and the goat anti-mouse secondary antibody (no. A-21058) were provided by Invitrogen. All other chemicals were from commercial sources.

Cells culture

A primary culture of human SMCs was established by explant outgrowth of a segment of human umbilical cord retrieved at the time of caesarean section (22). Endothelial cells were removed by scraping the luminal surface of the vessel with a cotton swab, and the adventitia was mechanically stripped away. Primary cultures were maintained in DMEM supplemented with 20% FBS and 1% antibiotics (penicillin/streptomycin) (all from Gibco). Cells between 4th and 10th passages were used in these experiments. The trail confirmed with the principle of the Declaration of Helsinki and was approved by the Ethics Committee of the Shanghai Ninth Hospital.

siRNA interference

β-catenin expression was inhibited by transfection with a siRNA specific to β-catenin. β-catenin siRNA was transiently transfected into the cells using Lipofectamine® 2000 (Invitrogen Life Technologies), according to the manufacturer's instructions. Briefly, 5×105 HUSMCs per well were cultured in 6-well plates to 75% confluence. The cells were then transfected with 100 pmol siRNA duplexes using 5 µl Lipofectamine® 2000 and 500 µl DMEM (reduced serum medium). The process of transfected was without antibiotics. Following a 72-h incubation at 37°C, the cells were harvested for analysis. The human β-catenin siRNA sequence was 5-CAT GUG UTG GUA AGC UCUA-3 and the scrambled siRNA sequence was 5-GCA ACA GTT GCA GAG AGGU-3. They were synthesized by Biotend (Shanghai, China).

Cell proliferation assay

Cell proliferation was measured with the CCK-8 assay kit. In brief, 5,000 cells were plated in each well of a 96-well plate and allowed to attach for 24 h. Then, HUSMCs were subsequently incubated for 0, 12, 24 or 48 h in the presence of gal-3. Subsequently, the plate was incubated with CCK-8 for 4 h at 37°C. At last, the absorbance of wave length was taken at 450 nm.

Migration assay

The assay was performed as previously described (23). Migration assays were performed in a chamber, HUSMCs were resuspended in 200 µl serum-free DMEM medium and 5×104 HUSMCs were loaded into the upper chambers. The lower chamber was filled with 400 µl of DMEM in the presence or absence of 10 µg/ml recombinant gal-3. The chamber was incubated at 37°C for 24 h. Then, the lower side of the filter was washed with PBS and fixed with 4% paraformaldeyde. Nuclei were stained with 4′,6-diamino-2-phenylindole (DAPI, 1:1,000; Sigma) for 5 min at room temperature. The cells were counted in three random high-power fields (x100) in each well.

Real-time RT-PCR analysis

Total RNA was extracted using TRIzol reagent (Invitrogen) according to the manufacturer's protocol. Total RNA was reverse-transcribed into cDNA using reverse transcriptase, and quantitative real-time PCR was performed using SYBR-Green master mix, on an Applied Biosystems 7500 real-time PCR system, according to the manufacturer's instructions. Specific primers for human gal-3, β-catenin, OPN, axin2, calponin, SMA and GAPDH were as follows: Gal-3 forward, 5′-GGCCACTGATTGTGCCTTAT-3′ and reverse, 5′-TGCAACCTTGAAGTGGTCAG-3′; β-catenin forward, 5′-GCCGGCTATTGTAGAAGCTG-3′ and reverse, 5′-GAGTCCCAAGGAGACCTTCC-3′; OPN forward, 5′-TGAGTCTGGAAATAACTAATGTGTTTGA-3′ and reverse, 5′-GAACATAGACATAACCCTGAAGCTTTT-3′; axin2 forward, 5′-CTCTCTACCTCATTTCCCGAGAAC-3′ and reverse, 5′-CGAGATCAGCTCAGCTGCAA-3′; calponin forward, 5′-ATGTGAGGAGGGAAGAGTGTG-3′ and reverse, 5′-CGGTTGAAGTGAGCAGAGG-3′; SMA forward, 5′-AGCGTGGCTACTCCTTCGTGAC-3′ and reverse, 5′-GCTCGTTGCCGATGGTGATGAC-3′; cyclin D1 forward, 5′-AATGACCCCGCACGATTTC-3′ and reverse, 5′-TCAGGTTCAGGCCTTGCAC-3′; GAPDH forward, 5′-TGATGACATCAAGAAGGTGGTGAAG-3′ and reverse, 5′-TCCTTGGAGGCCATGTGGGCCAT-3′.

Western blot analysis

Cells were lysed in a lysis buffer containing 150 mM of NaCl, 10 mM of Tris (pH 7.5), 5 mM of EDTA, 1% Triton X-100, 1 mM of PMSF, 10 mg/ml of leupeptin, 10 mg/ml of pepstatin, and 10 mg/ml of aprotinin for 30 min on ice. The nuclear and cytoplastic protein were separately extracted by using the kit (P 0028; Beyotime Institute of Biotechnology (Shanghai, China). Protein concentrations were measured with the BCA Protein Assay (Pierce Biotechnology Inc., Rockford, IL, USA). The lysates (20 µg) were electrophoresed on 10% SDS-PAGE and transferred to nitrocellulose membranes (Merck Millipore, Danvers, MA, USA). The membrane was blocked with 5% nonfat dry milk in TBST buffer (100 mM NaCl, 10 mM Tris-HCl, pH 7.4, and 0.1% Tween-20) for 1 h at room temperature. The blots were then incubated with various 1000-fold diluted primary antibodies at a dilution of 1:1,000 in TBST at 4°C overnight, and then washed twice with TBST buffer at room temperature and incubated for 1 h with the appropriate peroxidase-conjugated secondary antibody (1:5,000 dilution). All signals were detected by Odyssey (LI-COR Biosciences, Lincoln, NE, USA). To quantity the protein, band intensity was assessed by Quantity One 4.6.2 software.

Statistical analysis

All data are expressed as the mean ± SD. Statistics were performed using the SPSS 13.0 software (SPSS, Inc., Chicago, IL, USA). One-way ANOVA followed by the Student-Newman-Keuls post hoc analyses were used. For data that not passed the normality test; non-paramatric ANOVA (Kruskal-Wallis test) was used. A value of P<0.05 was considered statistically significant. All experiments were performed at least three times.

Results

Exogenous galectin-3 promotes HUSMCs proliferation and migration

First, we used increasing concentrations of gal-3 (up to 25 µg/ml) to stimulate cells. Cell proliferation was determined at 12, 24, and 48 h using an CCK-8 assay. As shown in Fig. 1A, the proliferation of HUSMCs was significantly increased in a concentration dependent manner. The Transwell migration assay was used to investigate the effect of recombined gal-3 on cell migration. As shown in Fig. 1B and C, exogenous gal-3 enhanced HUSMCs migration significantly.

Galectin-3 promotes the phenotype transformation of vascular smooth muscle cells

We also investigated the role of exogenous gal-3 in the phenotype transformation of HUSMC. First of all, the HUSMCs were cultured in the DMEM without serum for 12 h. After that, the serum-starved HUSMCs were treated with gal-3 for 48 h. As shown in the Fig. 2, the expression of the smooth muscle synthetic proteins, OPN rose significantly over a range of gal-3 concentration (0–25 µg/ml). Concomitant with the increased OPN expression, the protein markers of contractile phenotype were also increased in gal-3-treated cells, as measured by SMA and calponin expression.

Galectin-3 activates canonical Wnt signaling in human umbilical vascular smooth muscle cells

Canonical Wnt signal activation could be evaluated by recording β-catenin protein levels (24). The expression of β-catenin was tested by western blotting after cells were treated with gal-3. The distribution of β-catenin in cytoplasm and nucleu were examined by western blot analysis seperately. Gal3 obviousely increased the expression of β-catenin in nucleu, but it has relatively little effect on the cytoplasmic β-catenin. To further confirm the activation of canonical Wnt signaling by gal-3, we also detected the expression of axin2 (regulating β-catenin stability) (25) and cyclin D1 (β-catenin target gene) by using realtime-PCR and western blotting. Our results showed that recombinant gal-3 significantly increased expression of axin2 and cyclin D1 (Fig. 3). Gal-3 induced a marked increase in the protein expression of nonphosphorylated (active) and total β-catenin However, the mRNA level of β-catenin was almost unchanged in the gal-3-treated cells (Fig. 3C). In order to confirm that β-catenin expression was modulated by gal-3 at the post-translational level, we also detected the activity of GSK-3β which promotes the degradation of β-catenin. Phosphorylation GSK-3β was 2.5-fold increased by gal-3, while total GSK-3β were almostly unchanged.

Canonical Wnt/β-catenin signaling mediates the effects of galectin-3 in HUSMCs

To further evaluate the role of β-catenin in gal-3-mediated change of HUSMC, the expression of β-catenin was inhibited by siRNA strategy in HUSMCs. The siRNA of β-catenin reduced the expression of β-catenin mRNA and protein by 91 and 83%, respectively. These results indicated that it effectively antagonized the activation of Wnt/β-catenin signaling pathway. We then preformed cell proliferation and migration assays on these cells, the silencing of β-catenin decreased the proliferation and migration of HUSMCs (Fig. 1B and D). Furthermore, the increased ability of migration and proliferation in gal-3-treated cells were also eliminated after silencing of β-catenin. We then detected the role of β-catenin in the phenotypic switch induced by gal-3. β-catenin knockdown efficiently blocked the increase expression of OPN, SMA and calponin induced by gal-3 in both mRNA and protein levels, as demonstrated by western blot and realtime PCR. In order to further confirm the role the Wnt signaling pathway in gal-3 induced phenotype transformation of VSMC, XAV939, a inhibitor of Wnt signaling pathway, was also used. XAV939 similarly attenuated the effect of gal-3 in HUSMCs (Fig. 4A-C).

Discussion

In this study, we demonstrate, for the first time, that gal-3 can activate the canonical Wnt signaling pathway in HUSMCs. More importantly, we find that gal-3 promote the phenotype transformation of HUSMCs, including increasing proliferation and migration and upregulating a series of phenotype related proteins. While using the strategy of β-catenin siRNA can obviousely block the gal-3 induces phenotype transformation of HUSMCs. Taken all these data together, we conclude that gal-3 promotes the activation and phenotype transformation of HUSMCs through canonical Wnt/β-catenin signaling.

Gal-3 has been reported to affect the function of almost all the cells in the wall of blood vessel. In human umbilical vein endothelial cells (HUVEC), gal-3 is a mediator of vascular endothelial growth factor (VEGF)- and basic fibroblast growth factor (bFGF)-mediated angiogenesis (6,26). Gal-3 can also induce the migration of macrophage in mouse (27). Atherosclerosis is characterized by the activation and accumulation of smooth muscle cells in the intimal layer of blood vessels where they internalized a lot of lipids (28). The enhanced proliferation and migration of smooth muscle cells are the symbol of the early pathology of atherosclerosis. In this study, we also showed that exogenous gal-3 can obviously promote the proliferation and migration of HUSMCs. In order to further explore the role of gal-3 in the HUSMCs, we also detected the expression of some widely used SMC marker proteins.

Traditionally, smooth muscle cells are thought of being exist in two totally different phenotypes: Contractile and synthetic phenotype. It is now being recognized that there are a diversity of SMC phenotypes, ranging from contractile to synthetic (4,29,30). In fact, these so-called contractile and synthetic phenotypes should be envisaged as ‘idealized’ phenotypes. Higher growth rates and stronger migratory activity are usually considered typical of the ‘synthetic’ phenotype, and OPN is also identified as a synthetic-related protein (3). Actually, recent studies proved that different synthetic and contractile markers could be upregulated at the same time (4,26). In some instances, contractile differentiation can be up-regulated in the ‘synthetic’ phenotype and contractile differentiation markers may express with matrix synthesis (28,31). In our study, we found that gal-3 stimulated the expression of OPN. Interesting, at the same time, gal-3 also increased the contractile-related protein, calponin and SMA. Therefore, we believe that galectin-3 can promote expression of both smooth muscle synthetic and contractile proteins.

There are two different subclasses of Wnt signalling: The non-canonical pathway (which is β-catenin dependent) and the canonical pathway (β-catenin independent). The activation of non-canonical Wnt pathway can involve the intra-cytoplasmic release of Ca2+ and the activation of Jun N-terminal kinase (JNK). The canonical Wnt signalling pathway involves the inhibition of β-catenin degradation complex, therefore resulting in the translocation of β-catenin from the cytoplasm to the nucleus, and binding to downstream target genes. Earlier reports have shown that β-catenin can regulate the expression of OPN, SMA and calponin expression, induce cell proliferation and migration in many types of cells (28,32). Recently, the role of gal-3 in vascular smooth muscle cells osteogenic differentiation has also been confirmed (32). Thus, we explored the role of β-catenin in gal-3-treated HUSMCs. In our study, gal-3 activated the canonical Wnt signaling pathway by upregulating the total and nonphosphorylated β-catenin. We also found that the expression of β-catenin downstream gene axin2 and cyclin D1 was upregulated by gal-3 (28,32). It further indicated that Wnt/β-catenin signaling pathway was activated by gal-3 in the HUSMCs. Our results showed that gal-3 induced the expression of total β-catenin at protein level but not mRNA level. It is consensus with previous findings. Gal-3, a binding of β-catenin, increases phosphorylation of GSK-3β (12,33,34). Inactivation of GSK-3β can reduce the degradation of β-catenin, and increase cellular β-catenin levels besides, we found that gal-3 indeed increased the phosphorylated level of GSK-3β. In order to prove the role of β-catenin in gal-3-induced phenotype transformation of HUSMCs, we used the siRNA strategy and XAV939 to decrease the expression of β-catenin. Our results demonstrated that silencing of β-catenin inhibited gal-3-induced protein expression and blocked gal-3-mediated cell proliferation and migration of vascular smooth muscle cells. Thus, we conclude that recombinant gal-3 induces phenotype transformation of HUSMCs via canonical Wnt/β-catenin pathway.

In conclusion, our results show that gal-3 promotes the activation and the phenotype transformation in HUSMCs. This activity is through the canonical Wnt signaling pathway. Recently, gal-3 has been proved to be related to the atherosclerosis and coronary heart disease. As smooth muscle cells plays an important role in a variety of plausible mechanisms of atherosclerosis and chronic heart diseases. Our research could help to clarify the role of gal-3 in the pathological process of atherosclerosis.

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (81270376); Shanghai Hospital Development Center (SHDC12012312); Ministry of Education Science and Technology Development Center (20130073110016); Shanghai Science and Technology Commission (Chinese medicine) (12401905200) and the Fund of Ninth People's Hospital (grant no. 2013A02).

References

1 

Lusis AJ: Atherosclerosis. Nature. 407:233–241. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Karagiannis GS, Weile J, Bader GD and Minta J: Integrative pathway dissection of molecular mechanisms of moxLDL-induced vascular smooth muscle phenotype transformation. BMC Cardiovasc Disord. 13:42013. View Article : Google Scholar : PubMed/NCBI

3 

Doran AC, Meller N and McNamara CA: Role of smooth muscle cells in the initiation and early progression of atherosclerosis. Arterioscler Thromb Vasc Biol. 28:812–819. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Hao H, Gabbiani G and Bochaton-Piallat ML: Arterial smooth muscle cell heterogeneity: Implications for atherosclerosis and restenosis development. Arterioscler Thromb Vasc Biol. 23:1510–1520. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Kim K, Mayer EP and Nachtigal M: Galectin-3 expression in macrophages is signaled by Ras/MAP kinase pathway and up-regulated by modified lipoproteins. Biochim Biophys Acta. 1641:13–23. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Markowska AI, Liu FT and Panjwani N: Galectin-3 is an important mediator of VEGF- and bFGF-mediated angiogenic response. J Exp Med. 207:1981–1993. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Calvier L, Miana M, Reboul P, Cachofeiro V, Martinez-Martinez E, de Boer RA, Poirier F, Lacolley P, Zannad F, Rossignol P and López-Andrés N: Galectin-3 mediates aldosterone-induced vascular fibrosis. Arterioscler Thromb Vasc Biol. 33:67–75. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Dumic J, Dabelic S and Flögel M: Galectin-3: An open-ended story. Biochim Biophys Acta. 1760:616–635. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Shimura T, Takenaka Y, Tsutsumi S, Hogan V, Kikuchi A and Raz A: Galectin-3, a novel binding partner of beta-catenin. Cancer Res. 64:6363–6367. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Shimura T, Takenaka Y, Fukumori T, Tsutsumi S, Okada K, Hogan V, Kikuchi A, Kuwano H and Raz A: Implication of galectin-3 in Wnt signaling. Cancer Res. 65:3535–3537. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Song S, Mazurek N, Liu C, Sun Y, Ding QQ, Liu K, Hung MC and Bresalier RS: Galectin-3 mediates nuclear beta-catenin accumulation and Wnt signaling in human colon cancer cells by regulation of glycogen synthase kinase-3beta activity. Cancer Res. 69:1343–1349. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Francia P, Adduci C, Semprini L, Borro M, Ricotta A, Sensini I, Santini D, Caprinozzi M, Balla C, Simmaco M and Volpe M: Osteopontin and galectin-3 predict the risk of ventricular tachycardia and fibrillation in heart failure patients with implantable defibrillators. J Cardiovasc Electrophysiol. 25:609–616. 2014. View Article : Google Scholar : PubMed/NCBI

13 

de Boer RA, Edelmann F, Cohen-Solal A, Mamas MA, Maisel A and Pieske B: Galectin-3 in heart failure with preserved ejection fraction. Eur J Heart Fail. 15:1095–1101. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Felker GM, Fiuzat M, Shaw LK, Clare R, Whellan DJ, Bettari L, Shirolkar SC, Donahue M, Kitzman DW, Zannad F, et al: Galectin-3 in ambulatory patients with heart failure: Results from the HF-ACTION study. Circ Heart Fail. 5:72–78. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Mayr A, Klug G, Mair J, Streil K, Harrasser B, Feistritzer HJ, Jaschke W, Schocke M, Pachinger O and Metzler B: Galectin-3: Relation to infarct scar and left ventricular function after myocardial infarction. Int J Cardiol. 163:335–337. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Sanchez-Mas J, Lax A, Asensio-Lopez MC, Fernandez-Del Palacio MJ, Caballero L, Garrido IP, Pastor F, Januzzi JL and Pascual-Figal DA: Galectin-3 expression in cardiac remodeling after myocardial infarction. Int J Cardiol. 172:e98–e101. 2014. View Article : Google Scholar : PubMed/NCBI

17 

van Kimmenade RR, Januzzi JL Jr, Ellinor PT, Sharma UC, Bakker JA, Low AF, Martinez A, Crijns HJ, MacRae CA, Menheere PP and Pinto YM: Utility of amino-terminal pro-brain natriuretic peptide, galectin-3, and apelin for the evaluation of patients with acute heart failure. J Am Coll Cardiol. 48:1217–1224. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Szadkowska I, Wlazel RN, Migala M, Bajon-Laskowska K, Szadkowski K, Zielińska M, Paradowski M and Pawlicki L: The association between galectin-3 and occurrence of reinfarction early after first myocardial infarction treated invasively. Biomarkers. 18:655–659. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Nachtigal M, Ghaffar A and Mayer EP: Galectin-3 gene inactivation reduces atherosclerotic lesions and adventitial inflammation in ApoE-deficient mice. Am J Pathol. 172:247–255. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Seferovic JP, Lalic NM, Floridi F, Tesic M, Seferovic PM, Giga V, Lalic K, Jotic A, Jovicic S, Colak E, et al: Structural myocardial alterations in diabetes and hypertension: The role of galectin-3. Clin Chem Lab Med. 52:1499–1505. 2014.PubMed/NCBI

21 

Yilmaz H, Cakmak M, Inan O, Darcin T and Akcay A: Increased levels of galectin-3 were associated with prediabetes and diabetes: New risk factor? J Endocrinol Invest. 38:527–533. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Guyton JR, Lenz ML, Mathews B, Hughes H, Karsan D, Selinger E and Smith CV: Toxicity of oxidized low density lipoproteins for vascular smooth muscle cells and partial protection by antioxidants. Atherosclerosis. 118:237–249. 1995. View Article : Google Scholar : PubMed/NCBI

23 

Leavesley DI, Schwartz MA, Rosenfeld M and Cheresh DA: Integrin beta 1- and beta 3-mediated endothelial cell migration is triggered through distinct signaling mechanisms. J Cell Biol. 121:163–170. 1993. View Article : Google Scholar : PubMed/NCBI

24 

Tickenbrock L, Schwäble J, Strey A, Sargin B, Hehn S, Baas M, Choudhary C, Gerke V, Berdel WE, Müller-Tidow C and Serve H: Wnt signaling regulates transendothelial migration of monocytes. J Leukoc Biol. 79:1306–1313. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Schaale K, Neumann J, Schneider D, Ehlers S and Reiling N: Wnt signaling in macrophages: Augmenting and inhibiting mycobacteria-induced inflammatory responses. Eur J Cell Biol. 90:553–559. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Nangia-Makker P, Honjo Y, Sarvis R, Akahani S, Hogan V, Pienta KJ and Raz A: Galectin-3 induces endothelial cell morphogenesis and angiogenesis. Am J Pathol. 156:899–909. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Jia W, Kidoya H, Yamakawa D, Naito H and Takakura N: Galectin-3 accelerates M2 macrophage infiltration and angiogenesis in tumors. Am J Pathol. 182:1821–1831. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Carthy JM, Luo Z and McManus BM: WNT3A induces a contractile and secretory phenotype in cultured vascular smooth muscle cells that is associated with increased gap junction communication. Lab Invest. 92:246–255. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Rensen SS, Doevendans PA and van Eys GJ: Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth Heart J. 15:100–108. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Matsushita T, Rama A, Charolidi N, Dupont E and Severs NJ: Relationship of connexin43 expression to phenotypic modulation in cultured human aortic smooth muscle cells. Eur J Cell Biol. 86:617–628. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Rama A, Matsushita T, Charolidi N, Rothery S, Dupont E and Severs NJ: Up-regulation of connexin43 correlates with increased synthetic activity and enhanced contractile differentiation in TGF-beta-treated human aortic smooth muscle cells. Eur J Cell Biol. 85:375–386. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Menini S, Iacobini C, Ricci C, Fantauzzi C Blasetti, Salvi L, Pesce CM, Relucenti M, Familiari G, Taurino M and Pugliese G: The galectin-3/RAGE dyad modulates vascular osteogenesis in atherosclerosis. Cardiovasc Res. 100:472–480. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Kobayashi T, Shimura T, Yajima T, Kubo N, Araki K, Tsutsumi S, Suzuki H, Kuwano H and Raz A: Transient gene silencing of galectin-3 suppresses pancreatic cancer cell migration and invasion through degradation of β-catenin. Int J Cancer. 129:2775–2786. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Zhang D, Chen ZG, Liu SH, Dong ZQ, Dalin M, Bao SS, Hu YW and Wei FC: Galectin-3 gene silencing inhibits migration and invasion of human tongue cancer cells in vitro via downregulating β-catenin. Acta Pharmacol Sin. 34:176–184. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2017
Volume 15 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tian L, Chen K, Cao J, Han Z, Wang Y, Gao L, Fan Y and Wang C: Galectin‑3 induces the phenotype transformation of human vascular smooth muscle cells via the canonical Wnt signaling. Mol Med Rep 15: 3840-3846, 2017
APA
Tian, L., Chen, K., Cao, J., Han, Z., Wang, Y., Gao, L. ... Wang, C. (2017). Galectin‑3 induces the phenotype transformation of human vascular smooth muscle cells via the canonical Wnt signaling. Molecular Medicine Reports, 15, 3840-3846. https://doi.org/10.3892/mmr.2017.6429
MLA
Tian, L., Chen, K., Cao, J., Han, Z., Wang, Y., Gao, L., Fan, Y., Wang, C."Galectin‑3 induces the phenotype transformation of human vascular smooth muscle cells via the canonical Wnt signaling". Molecular Medicine Reports 15.6 (2017): 3840-3846.
Chicago
Tian, L., Chen, K., Cao, J., Han, Z., Wang, Y., Gao, L., Fan, Y., Wang, C."Galectin‑3 induces the phenotype transformation of human vascular smooth muscle cells via the canonical Wnt signaling". Molecular Medicine Reports 15, no. 6 (2017): 3840-3846. https://doi.org/10.3892/mmr.2017.6429